Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Biol Rep ; 51(1): 118, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38227082

RESUMO

BACKGROUND: Ovarian cancer is one of the most lethal gynecological cancers among women worldwide. Cisplatin (Cis) is an effective chemotherapeutic agent used to treat several types of cancer. Silymarin (SLM) is an extract of medicinal plant Silybum marianum (milk thistle) with anti-inflammatory, anti-angiogenesis, antioxidant, and anticancer properties used alone or in combination with other drugs. OBJECTIVE: This study aimed to explore the effects of co-treatment with SLM and Cis on A2780 human ovarian cancer cell lines. METHODS: In this study, A2780 cells were treated with various concentrations of SLM and Cis, separately and in combination. Cell cytotoxicity, scratch, clonogenic, and flow-cytometry assays were accomplished to estimate cell viability, migration, colony formation, and apoptosis, respectively. Real-time PCR was utilized to determine the expression levels of miR-155 and miR-27a. RESULTS: SLM significantly reduced the proliferation of A2780 cells in a concentration- and time-dependent manner. Combination treatment with SLM and Cis was more potent than either single treatment in reducing viability, suppressing migration, inhibiting colony formation, and promoting the induction of apoptosis. Additionally, gene expression analysis revealed a significant decline in the expression levels of miR-155 and miR-27a in response to all separate and combined treatments, and co-treatment was more effective than individual treatments in altering miRNAs expression. CONCLUSION: Based on our findings, SLM boosts the anticancer activity of Cis and mitigates its side effects. Thus, the co-treatment of SLM and Cis can be proposed as a promising therapeutic strategy for further investigation.


Assuntos
MicroRNAs , Neoplasias Ovarianas , Silimarina , Feminino , Humanos , Cisplatino/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Silimarina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Apoptose , MicroRNAs/genética
2.
Toxicol In Vitro ; 93: 105687, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37659683

RESUMO

This study aimed to investigate the effects of glycyrrhizic acid (GL) on the anticancer activity of cisplatin in A2780 ovarian cancer cells. Cultured A2780 cells were treated with different concentrations of GL and cisplatin individually and in combination. The MTT assay, flow cytometry, wound-healing, and clonogenic assay, were used to determine cell viability, apoptosis, migration, and colony formation, respectively. The effects on superoxide dismutase (SOD) activity were also evaluated. QPCR was used to study the effects of individual and combined treatments with GL and cisplatin on the expression levels of migration genes (MMP2 and MMP9), and some apoptosis pathway genes (caspase-3, -8, -9, and BCL2). A synergistic effect was observed between GL and cisplatin (CI < 1). Combination therapy was significantly more effective in reducing cell viability, suppressing migration and colony formation, inducing apoptosis, and altering gene expression compared to single therapies. GL significantly increased SOD activity. The relative expression of caspase -3, -8, and - 9 increased significantly, and the expression levels of MMP2 and MMP9 decreased significantly in the treated cells. Our results indicate that GL enhances the anticancer activity of cisplatin in the A2780 cell line. Therefore, the combination of GL and cisplatin can be proposed as a promising therapeutic strategy for ovarian cancer.


Assuntos
Antineoplásicos , Neoplasias Ovarianas , Humanos , Feminino , Cisplatino/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Ácido Glicirrízico/farmacologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Apoptose , Superóxido Dismutase/genética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Proliferação de Células
3.
Immunotherapy ; 15(14): 1157-1169, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37584216

RESUMO

Aim: We predicted the modulation of autophagy and apoptosis in response to temozolomide (TMZ) and IFN-γ based on changes in the expression of non-coding RNAs in C6-induced glioblastoma (GBM). Materials & methods: Each rat received an intraperitoneal injection of TMZ (7.5 mg/kg) and/or IFN-γ (50,000 IU). Results: The reduced expression of H19 and colorectal neoplasia differentially expressed (CRNDE) was associated with a reduction in autophagy in response to TMZ, IFN-γ and TMZ + IFN-γ therapy, whereas the decreased level of miR-29a (proapoptotic miRNA) was associated with an increase in apoptosis. Conclusion: It appears that H19 promotes switching from autophagy to apoptosis in response to combination therapy of TMZ and IFN-γ through the miR-29a/autophagy-related protein 9A (ATG9A) pathway in C6-induced GBM.


Temozolomide (TMZ) is a drug for people with brain cancer. It can make it hard for patients to learn and think, and it can also make the drug stop working, which lets the tumor keep growing. Researchers are looking for other drugs or things that can be taken with TMZ to stop this from happening. In this study, we used a protein called interferon (IFN), which helps fight cancer. We gave mice with brain cancer both TMZ and IFN, and saw that the tumor cells died and the tumor got smaller. We also looked at how IFN and TMZ changed the genetic material of the mouse brain, called RNA. But we need to test this on people to be sure it works.


Assuntos
Neoplasias Encefálicas , Glioblastoma , MicroRNAs , Ratos , Animais , Temozolomida/uso terapêutico , Temozolomida/farmacologia , Glioblastoma/tratamento farmacológico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , MicroRNAs/genética , Autofagia , Apoptose , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico
4.
J Mol Neurosci ; 71(12): 2566-2574, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34041686

RESUMO

Cohen syndrome is caused by homozygous mutation in the vacuolar protein sorting 13 homolog B (VPS13B, also referred to as COH1) gene on chromosome 8q22.2. The VPS13B protein is involved in transmembrane transport, Golgi integrity, and neuritogenesis. Clinical manifestations of Cohen syndrome are mainly intellectual disability, developmental delay, facial abnormalities, and eye disorders. This study aimed to identify the causative variant in two unrelated families with Cohen syndrome. To this end, whole-exome sequencing (WES) was performed to identify the pathogenic variants. A homozygous nonsense variant (NM_017890:c.10369C > T; NP_060360.3: p.Q3457X) in the VPS13B gene was identified and co-segregated with all affected individuals in both families. In silico analysis highly suggested this variant as damaging for protein function. The present study increases the mutation spectrum of the VPS13B gene and could be useful in genetic diagnosis and genetic counseling in Cohen syndrome patients.


Assuntos
Dedos/anormalidades , Deficiência Intelectual/genética , Microcefalia/genética , Hipotonia Muscular/genética , Miopia/genética , Obesidade/genética , Degeneração Retiniana/genética , Proteínas de Transporte Vesicular/genética , Criança , Códon sem Sentido , Deficiências do Desenvolvimento/genética , Deficiências do Desenvolvimento/patologia , Dedos/patologia , Homozigoto , Humanos , Deficiência Intelectual/patologia , Masculino , Microcefalia/patologia , Hipotonia Muscular/patologia , Miopia/patologia , Obesidade/patologia , Fenótipo , Degeneração Retiniana/patologia , Sequenciamento do Exoma , Adulto Jovem
5.
J Immunother Cancer ; 9(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33472857

RESUMO

Exosomes, as the main group of extracellular vesicles, are biologically active lipid-bilayer vesicles that are naturally released from different types of normal or tumor cells. These vesicles play an important role in intercellular communication and influence the extracellular environment and the immune system. Emerging evidence demonstrates that cancer-derived exosomes are enriched in immunosuppressive proteins, such as the programmed death-ligand 1 (PD-L1). PD-L1 and its receptor programmed cell death protein 1 (PD-1) are the key immune checkpoint molecules that promote tumor progression via negative regulation of immune responses. PDL-1 is highly expressed on the surface of tumor cells and binds to PD-1 on the surface of activated T cells, leading to suppression of T cells, which consequently enables cancer cells to escape antitumor immunity. Currently, there are several Food and Drug Administration-approved monoclonal antibodies blocking PD-1/PD-L1 interaction, which are clinically used for cancer treatment. However, despite impressive treatment outcomes, some patients show poor response to PD-1/PD-L1 blockade. Of note, tumor-derived exosomes containing PD-L1 can recapitulate the effect of cell-surface PD-L1. There is evidence that reveals a significant association between levels of circulating exosomal PD-L1 and rate of response to anti-PD-1/PD-L1 antibody therapy. The present article reviews the role of exosomal PDL-1 in the therapeutic resistance to anti-PD-1/PD-L1 treatment. Importantly, it is suggested that the removal of exosomal PDL-1 could serve as a therapeutic adjuvant for enhancing the efficacy of anti-PD-1/PD-L1 therapy in patients with cancer.


Assuntos
Antígeno B7-H1/metabolismo , Resistencia a Medicamentos Antineoplásicos , Exossomos/metabolismo , Neoplasias/imunologia , Antígeno B7-H1/efeitos dos fármacos , Progressão da Doença , Exossomos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/tratamento farmacológico , Receptor de Morte Celular Programada 1/efeitos dos fármacos , Receptor de Morte Celular Programada 1/metabolismo , Regulação para Cima/efeitos dos fármacos
6.
Cancer Gene Ther ; 28(3-4): 157-174, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32773776

RESUMO

Disruption in DNA methylation processes can lead to alteration in gene expression and function that would ultimately result in malignant transformation. In this way, studies have shown that, in cancers, methylation-associated silencing inactivates tumor suppressor genes, as effectively as mutations. DNA methylation machinery is composed of several genes, including those with DNA methyltransferases activity, proteins that bind to methylated cytosine in the promoter region, and enzymes with demethylase activity. Based on a prominent body of evidence, DNA methylation machinery could be regulated by microRNAs (miRNAs) called epi-miRNAs. Numerous studies demonstrated that dysregulation in DNA methylation regulators like upstream epi-miRNAs is indispensable for carcinogenesis; consequently, the malignant capacity of these cells could be reversed by restoring of this regulatory system in cancer. Conceivably, recognition of these epi-miRNAs in cancer cells could not only reveal novel molecular entities in carcinogenesis, but also render promising targets for cancer therapy. In this review, at first, we have an overview of the methylation alteration in cancers, and the effect of this phenomenon in miRNAs expression and after that, we conduct an in-depth discussion about the regulation of DNA methylation regulators by epi-miRNAs in cancer cells.


Assuntos
MicroRNAs/genética , Neoplasias/terapia , Metilação de DNA , Humanos , Neoplasias/genética
7.
J Oncol ; 2020: 7630197, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33224198

RESUMO

Lung cancer is the most common cause of cancer-related deaths worldwide. Annually, millions of people die from lung cancer because of late detection and ineffective therapies. Recently, exosomes have been introduced as new therapeutic players with the potential to improve upon current diagnostic and treatment options. Exosomes are small membranous vesicles produced during endosomal merging. This allows for cell packaging of nucleic acids, proteins, and lipids and transfer to adjacent or distant cells. While exosomes are a part of normal intercellular signaling, they also allow malignant cells to transfer oncogenic material leading to tumor spread and metastasis. Exosomes are an interesting field of discovery for biomarkers and therapeutic targets. Among exosomal materials, lncRNAs have priority; lncRNAs are a class of noncoding RNAs longer than 200 base pairs. In the case of cancer, primary interest regards their oncogene and tumor suppressor functions. In this review, the advantages of exosomal lncRNAs as biomarkers and therapeutic targets will be discussed in addition to reviewing studies of their application in lung cancer.

8.
Int Rev Immunol ; 39(6): 264-279, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32552273

RESUMO

There is a high level of heterogeneity in symptom manifestations and response to disease-modifying therapies (DMTs) in multiple sclerosis (MS), an immune-based neurodegenerative disease with ever-increasing prevalence in recent decades. Because of unknown aspects of the etiopathology of MS and mechanism of action of DMTs, the reason for this variability is undetermined, and much remains to be understood. Traditionally, physicians consider switching to other DMTs based on the exacerbation of symptoms and/or change in the results of magnetic resonance imaging and biochemical factors. Therefore, identifying biological treatment response markers that help us recognizing non-responders rapidly and subsequently choosing another DMTs is necessary. microRNAs (miRNAs) are micromanagers of gene expression which have been profiled in different samples of MS patients, highlighting their role in pathogenetic of MS. Recent studies have investigated expression profiling of miRNAs after treatment with DMTs to clarify possible DMTs-mediated mechanism and obtaining response to therapy biomarkers. In this review, we will discuss the modulation of miRNAs by DMTs in cells and pathways involved in MS.


Assuntos
Regulação da Expressão Gênica , Fatores Imunológicos/uso terapêutico , MicroRNAs/genética , Esclerose Múltipla/genética , Esclerose Múltipla/terapia , Animais , Biomarcadores , Gerenciamento Clínico , Suscetibilidade a Doenças , Humanos , Fatores Imunológicos/farmacologia , Imunomodulação/efeitos dos fármacos , Esclerose Múltipla/diagnóstico , Esclerose Múltipla/imunologia , Resultado do Tratamento
9.
Cell Commun Signal ; 18(1): 88, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32517694

RESUMO

Autophagy has a crucial role in many cancers, including brain tumors. Several types of endogenous molecules (e.g. microRNAs, AKT, PTEN, p53, EGFR, and NF1) can modulate the process of autophagy. Recently miRNAs (small non-coding RNAs) have been found to play a vital role in the regulation of different cellular and molecular processes, such as autophagy. Deregulation of these molecules is associated with the development and progression of different pathological conditions, including brain tumors. It was found that miRNAs are epigenetic regulators, which influence the level of proteins coded by the targeted mRNAs with any modification of the genetic sequences. It has been revealed that various miRNAs (e.g., miR-7-1-3p, miR-340, miR-17, miR-30a, miR-224-3p, and miR-93), as epigenetic regulators, can modulate autophagy pathways within brain tumors. A deeper understanding of the underlying molecular targets of miRNAs, and their function in autophagy pathways could contribute to the development of new treatment methods for patients with brain tumors. In this review, we summarize the various miRNAs, which are involved in regulating autophagy in brain tumors. Moreover, we highlight the role of miRNAs in autophagy-related pathways in different cancers. Video abstract.


Assuntos
Autofagia , Neoplasias Encefálicas/metabolismo , MicroRNAs/fisiologia , Animais , Humanos
10.
Mol Biol Rep ; 47(5): 4099-4103, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32367316

RESUMO

Joubert syndrome (JS) disease is a clinically and genetically heterogeneous disorder with mutations in more than 35 genes involved in its pathogenicity. Molecular genetic methods including next generation sequencing (NGS) and Sanger sequencing are effective techniques used for identifying rare genetic variants that have a strong effect on disease pathogenesis. In this study, we tested a large pedigree with a history of several affected members with JS. At first the proband was sequenced by NGS technique then, confirmed by sanger sequencing method. After this, all available members of the pedigree were subjected to molecular analysis by sanger sequencing technique. The results of this study showed a novel variant in the C5ORF42 gene c.3080A > T: p. D1027V leading to a substitution of a valine for aspartic acid (D1027V) and may be associated with JS. This variant was present in proband compatible with autosomal recessive pattern. Also this variant was present in all parents (both father and mother) of affected individuals in a heterozygous state. It seems that mutations in C5ORF42 gene are associated with JS. However, the substantial mechanism requires further investigation.


Assuntos
Anormalidades Múltiplas/genética , Cerebelo/anormalidades , Anormalidades do Olho/genética , Doenças Renais Císticas/genética , Proteínas de Membrana/genética , Retina/anormalidades , Anormalidades Múltiplas/fisiopatologia , Adulto , Encéfalo/fisiologia , Cerebelo/fisiologia , Cerebelo/fisiopatologia , Pré-Escolar , Anormalidades do Olho/fisiopatologia , Feminino , Heterozigoto , Humanos , Lactente , Doenças Renais Císticas/fisiopatologia , Masculino , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Mutação , Linhagem , Retina/fisiopatologia , Sequenciamento do Exoma/métodos
11.
Cancer Gene Ther ; 27(12): 854-868, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32418986

RESUMO

Cancer immunotherapy emerged as a novel therapeutic option that employs enhanced or amended native immune system to create a robust response against malignant cells. The systemic therapies with immune-stimulating cytokines have resulted in substantial dose-limiting toxicities. Targeted cytokine immunotherapy is being explored to overcome the heterogeneity of malignant cells and tumor cell defense with a remarkable reduction of systemic side effects. Cell-based strategies, such as dendritic cells (DCs), fibroblasts or mesenchymal stem cells (MSCs) seek to minimize the numerous toxic side effects of systemic administration of cytokines for extended periods of time. The usual toxicities comprised of a vascular leak, hypotension, and respiratory insufficiency. Natural and strong tropism of MSCs toward malignant cells made them an ideal systemic delivery vehicle to direct the proposed therapeutic genes to the vicinity of a tumor where their expression could evoke an immune reaction against the tumor. Compared with other methods, the delivery of cytokines via engineered MSCs is safer and renders a more practical, and promising strategy. Large numbers of genes code for cytokines have been utilized to reengineer MSCs as therapeutic cells. This review highlights the recent findings on the cytokine gene therapy for human malignancies by focusing on MSCs application in cancer immunotherapy.


Assuntos
Engenharia Genética/métodos , Imunomodulação/imunologia , Imunoterapia/métodos , Células-Tronco Mesenquimais/metabolismo , Neoplasias/imunologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos
12.
Cancer Gene Ther ; 27(10-11): 739-753, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32203060

RESUMO

Inflammation, among environmental risk factors, is one of the most important contributors to colorectal cancer (CRC) development. In this way, studies revealed that the incidence of CRC in inflammatory bowel disease patients is up to 60% higher than the general population. MicroRNAs (miRNAs), small noncoding RNA molecules, have attracted excessive attention due to their fundamental role in various aspects of cellular biology, such as inflammation by binding to the 3'-untranslated regions (3'-UTR) of pro and anti-inflammatory genes. Based on multiple previous studies, SNPs at 3'-UTR can affect miRNA recognition elements by changing the thermodynamic features and secondary structure. This effect can be categorized, based on the number of changes, into four groups, including break, decrease, create, and enhance. In this paper, we will focus on functional variants in miRNA binding sites in inflammatory genes, which can modulate the risk of CRC by both investigating previous studies, regarding miRSNPs in inflammatory genes associated with CRC and recruiting in silico prediction algorithms to report putative miRSNPs in 176 inflammatory genes. In our analysis, we achieved 110 miRSNPs in 3'-UTR of 67 genes that seem good targets for future researches.


Assuntos
Neoplasias Colorretais/genética , Biologia Computacional/métodos , Predisposição Genética para Doença/genética , Polimorfismo Genético/genética , Polimorfismo de Nucleotídeo Único/genética , Feminino , Humanos , Masculino
13.
IUBMB Life ; 72(3): 343-360, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31889417

RESUMO

Cellular microRNAs (miRNAs) were identified as a key player in the posttranscriptional regulation of cellular-genes regulatory pathways. They also emerged as a significant regulator of the immune response. In particular, miR-146a acts as an importance modulator of function and differentiation cells of the innate and adaptive immunity. It has been associated with disorder including cancer and viral infections. Given its significance in the regulation of key cellular processes, it is not surprising which virus infection have found ways to dysregulation of miRNAs. miR-146a has been identified in exosomes (exosomal miR-146a). After the exosomes release from donor cells, they are taken up by the recipient cell and probably the exosomal miR-146a is able to modulate the antiviral response in the recipient cell and result in making them more susceptible to virus infection. In this review, we discuss recent reports regarding miR-146a expression levels, target genes, function, and contributing role in the pathogenesis of the viral infection and provide a clue to develop the new therapeutic and preventive strategies for viral disease in the future.


Assuntos
MicroRNAs/fisiologia , Viroses/genética , Exossomos/genética , Regulação da Expressão Gênica , Infecções por HIV/genética , Infecções por HIV/imunologia , Hepatite B/genética , Hepatite B/imunologia , Hepatite C/genética , Hepatite C/imunologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno/genética , Humanos , Influenza Humana/genética , Influenza Humana/imunologia , Viroses/imunologia
14.
Crit Rev Oncol Hematol ; 145: 102854, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31877535

RESUMO

Both environmental and genetic factors are involved in the initiation and development of gastrointestinal cancer. Covalent closed circular RNAs (circRNAs) are produced by a mechanism called "back-splicing" from mRNAs. They are highly stable and show cell and tissue specific expression patterns. Although some functions such as "microRNA sponge" and "RNA binding protein sponge" have been reported for a small number of circRNAs, the function of thousands of other circRNAs is still unknown. Dysregulation of circRNAs has been reported in many GI cancers and are involved in metastasis and invasion. CircRNAs have been reported to be useful as prognostic markers and targets for developing new treatments. We first describe the properties and biogenesis of circRNAs. We then summarize recent reports about circRNA functions, expression status, and their potential to be used as biomarkers in GI cancers including, gastric cancer, colorectal cancer, esophageal cancer, hepatocellular carcinoma, gallbladder cancer and pancreatic cancer.


Assuntos
Neoplasias Gastrointestinais , MicroRNAs , Splicing de RNA , RNA Circular , Biomarcadores , Epigênese Genética , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/patologia , Humanos , MicroRNAs/genética , Prognóstico
15.
Immunol Res ; 67(6): 505-516, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-32026322

RESUMO

It is believed that pathways of the immune system are responsible for eradicating cancer cells; however, their over-activation and also their ectopic expression in tumor cells and microenvironment are major contributors to tumor growth and chemoresistance. Toll-like receptor 4 (TLR4) pathway is an innate immune-related pathway which is usually overexpressed in tumor cells that leads to excessive pro-inflammatory cytokines and eventually results in tumor survival, drug resistance, and metastasis. In this study, we investigated whether TLR4 expression is affected upon the treatment of breast and ovarian cancer cells with common chemotherapeutics (paclitaxel, cisplatin, doxorubicin, and arsenic trioxide) and if TLR4 inhibition using its specific inhibitor TAK-242 could enhance cancer cells' response to the drugs. Both breast (MCF7) and ovarian (2008C13) cancer cells experienced an elevated expression of TLR4 after treatment with the drugs. The expression of this receptor was also upregulated in cisplatin-resistant 2008C13 cells; however, it was significantly higher upon short-term treatment with cisplatin. More importantly, the combination treatment of the drugs with TAK-242 intensified the chemosensitivity of six different breast and ovarian cancer cells to chemotherapeutic drugs. It was also identified that co-treatment of paclitaxel and TAK-242 not only led to enhanced G2/M arrest and apoptosis but also satisfactorily decreased the expression of TLR4 and different interleukins in these cells. Taken together, the results of the present study emphasize that chemotherapy may lead to chemoresistance through inducing TLR4 expression, and therefore inhibiting this receptor using TAK-242 could be a promising approach to improve the outcome of chemotherapy in foreseeable future.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Sulfonamidas/farmacologia , Receptor 4 Toll-Like/antagonistas & inibidores , Neoplasias da Mama/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Sinergismo Farmacológico , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Células MCF-7 , Neoplasias Ovarianas/metabolismo , Paclitaxel/farmacologia , Microambiente Tumoral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...